WJ E M

World Journal of Experimental Medicine World J Exp Med 2015 May 20; 5(2): 40-49 ISSN 2220-315X (online)

Submit a Manuscript: http://www.wjgnet.com/esps/ Help Desk: http://www.wjgnet.com/esps/helpdesk.aspx DOI: 10.5493/wjem.v5.i2.40

© 2015 Baishideng Publishing Group Inc. All rights reserved.

REVIEW

Endothelium-derived essential signals involved in pancreas organogenesis Dodanim Talavera-Adame, Donald C Dafoe differentiation, endocrine specification, and endocrine function. They are also involved in the physiopathology of type 1 and type 2 diabetes. During embryogenesis, aortic ECs provide specific factors that maintain the expression of key genes for pancreas development such as pancreatic and duodenal homeobox-1. Other unknown factors are also important for pancreatic endocrine specification and formation of insulin-producing beta cells. Endocrine precursors proliferate interspersed with ductal cells and exocrine precursors and, at some point of development, these endocrine precursors migrate to pancreatic mesenchyme and start forming the islets of Langerhans. By the end of the gestation and close to birth, these islets contain immature beta cells with the capacity to express vascular endothelial growth factor and therefore to recruit ECs from the surrounding microenvironment. ECs in turn produce factors that are essential to maintain insulin secretion in pancreatic beta cells. Once assembled, a cross talk between endocrine cells and ECs maintain the integrity of islets toward an adequate function during the whole life of the adult individual. This review will focus in the EC role in the differentiation and maturation of pancreatic beta cells during embryogenesis as well as the current knowledge about the involvement of endothelium to derive pancreatic beta cells in vitro from mouse or human pluripotent stem cells.

Dodanim Talavera-Adame, Donald C Dafoe, Comprehensive Transplant Center, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States Dodanim Talavera-Adame, Donald C Dafoe, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States Author contributions: Talavera-Adame D and Dafoe DC contributed equally to this work; Talavera-Adame D and Dafoe DC design the research, contributed new reagents/analytic tools, analyzed the data, and wrote the paper; Talavera-Adame D performed the research. Supported by Eris M. Field Endowment for Diabetes Research assigned to Donald C Dafoe. Conflict-of-interest: The authors have nothing to declare regarding any conflict of interests. Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/ licenses/by-nc/4.0/ Correspondence to: Donald C Dafoe, MD, Professor of Surgery, Clinical Professor of Surgery, UCLA, Eris M. Field Chair in Diabetes Research, Vice-Chair of Education, Director, Comprehensive Transplant Center, Department of Surgery, CedarsSinai Medical Center, 8635 W. Third Street, Suite 590W, Los Angeles, CA 90048, United States. [email protected] Telephone: +1-310-4237899 Fax: +1-310-4233864 Received: December 6, 2014 Peer-review started: December 6, 2014 First decision: January 20, 2015 Revised: March 18, 2015 Accepted: April 10, 2015 Article in press: April 12, 2015 Published online: May 20, 2015

Key words: Endothelium; Endothelium-derived signals; Differentiation; Pancreas development; Organogenesis © The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.

Core tip: Many studies have demonstrated that endothelial cells (ECs) have an important role in organogenesis. For instance, during embryogenesis, aortic ECs provide specific factors that maintain the expression of key genes for pancreas development. Other unknown factors are also important for pancreatic endocrine specification and formation of insulin-producing beta cells. In addition, by the end of the gestation and close to birth, pancreatic

Abstract Endothelial cells (ECs) are essential for pancreas

WJEM|www.wjgnet.com

40

May 20, 2015|Volume 5|Issue 2|

Talavera-Adame D et al . Endothelium role in pancreas organogenesis

islets contain immature beta cells with the capacity to express factors that recruit ECs from the surrounding microenvironment and form a functional unit that will lasts for the whole life of the individual. In the present review, we will analyze the current endothelial-derived factors called angiocrine factors that are essential in organogenesis and we will focus the role of these factors in pancreas development and pancreatic beta cells.

The term angiocrine has been proposed by Butler and collaborators to indicate the capacity of ECs to release growth factors and cytokines that may be involved in [25] organogenesis . For instance, bone marrow sinusoidal ECs (SECs) promote differentiation of hematopoietic stem cells (HSCs) through “angiocrine factors” such as [25,26] hepatocyte growth factor (HGF), Wnt2, and Notch . Additionally, Butler and colleagues reported that ECs promote self-renewal of hematopoietic stem cells when [25] these cells are interacting in co-culture . Apparently, Notch ligands expressed by ECs are associated with [25] this response . Another studies indicate that ECs from liver, called sinusoidal ECs (LSECs), express factors such as VE-cadherin, Factor VIII and vascular endothelial growth factors 2 and 3 (VEGF2, VEGF3) and these cells release angiocrine factors that may be involved [26] in liver regeneration . In another work it has been described that angiocrine factors are able to regulate [27] tumor growth . Other studies demonstrated that mice deficient in FLK-1 (VEGF receptor), die at embryonic day E9.5 or E10.5 because immaturity of blood cells and [28] blood vessels . Absence of embryonic liver budding is also present in these mice indicating that ECs play an important role during the early phases of organogenesis. In a similar work, it has been observed that ECs are also involved during the early stages of pancreas [20] development . Another research work indicated that signals from myocardium, such as those exerted by bone morphogenetic protein-2 (BMP-2), can promote epithelial-mesenchymal transformation mediated by [24,29] ECs . In the kidney, VEGF, bFGF, and PDGF coordinate [30] cellular differentiation, proliferation, and migration . It has been suggested that ECs promote the differentiation of endoderm cells toward liver or pancreas through [17,19,24,28,31,32] secretion of HGF (Table 1). At the same time, reciprocal interactions between tissue-specific cell types and ECs ensure coordinated growth and adequate tissue function. For instance, it is known that neurogenesis takes place close to blood vessels in adult [33] brains . Additionally, brain-derived neurotrophic factor (BDNF) secreted by ECs promotes neurogenesis and [34] angiogenesis in the brain of song birds . Another report indicates that pigment epithelium-derived factor (PEDF) is secreted by ECs and enhances self-renewal of neural [35] stem cells (NSCs) . Regarding the pancreas, it has been shown that pancreatic endoderm attract endothelial progenitor cells (EPCs) or angioblasts by expression [36] of SDF-1/CXCL12 . Expression of pancreatic and duodenal homeobox 1 (PDX-1) appeared in endoderm cells in contact with angioblasts via LIM domain only 2 (LMO2) suggesting that angioblasts may induce [37] expression of PDX-1 . Functional blood vessels may induce differentiation before they carry blood. However, some blood factors such as sphingosine-1-phosphate [38] (SIP) are important for differentiation and maturation . For instance, it has also been described that beta-cell differentiation can be regulated by oxygen tension via [39] hypoxia-inducible factor 1 alpha (HIF-1α) . This fact suggests that blood factors can also be involved in the

Talavera-Adame D, Dafoe DC. Endothelium-derived essential signals involved in pancreas organogenesis. World J Exp Med 2015; 5(2): 40-49 Available from: URL: http://www.wjgnet.com/ 2220-315X/full/v5/i2/40.htm DOI: http://dx.doi.org/10.5493/ wjem.v5.i2.40

INTRODUCTION At present, insulin producing cells have been derived from [1-3] different sources . With the emergence of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), that can be plated in vitro, the potential production of several cell types including pancreas [2,4-11] can be now achieved . Endothelial cells (ECs) are [12-15] . present at early stages during embry­ogenesis [16] These cells are the main component of most niches . Therefore, ECs interact early with developing tissues during organogenesis even before they are able to [16-19] form blood vessels and nourish specific regions . It has been reported that the absence of ECs results in [20,21] agenesis of some organs such as pancreas . This fact points out the essential role of EC signaling during organogenesis. Apparently ECs are not only involved in pancreas differentiation, they are essential for endocrine [20] differentiation as well . Endocrine progenitors give rise to immature beta cells that recruit ECs after expression [18,20] of vascular endothelial cell growth factor (VEGF) . These ECs in turn provide factors to promote beta-cell maturation and stabilize b-cell function. In this review, we will be focusing in the role of ECs in the differentiation and maturation of beta cells in vivo and in vitro with the emerging technology of human PSCs that can be expanded in vitro.

ENDOTHELIUM AS AN ENDOCRINE ORGAN Extensive studies with ECs have demonstrated that these cells play essential roles in immunity, inflammation, angiogenesis, and tumor metastasis. ECs do not only line [22,23] the interior surface of blood vessels . ECs are found at the interface between blood and other cell types. They not only maintain the blood fluid but also have a great plasticity that allows these cells to accomplish several [18] essential functions to maintain homeostasis . However, recent studies demonstrated that the endothelium is capable of releasing growth factors and cytokines that [24] play an essential role in organogenesis (Table 1).

WJEM|www.wjgnet.com

41

May 20, 2015|Volume 5|Issue 2|

Talavera-Adame D et al . Endothelium role in pancreas organogenesis Table 1 EC-derived factors related to organogenesis Angiocrine factors

Source

Notch ligands (1 and 2) VEGF bFGF PDGF HGF

Target

Ref

Bone marrow ECs; brain ECs ECs ECs Brain capillaries Lunga capillaries, SECs, islet capillaries

Neural progenitors and HSCs [40,41] ECs and several tissues such as islets [12,42,43] ECs and several tissues such as islets [43] ECs [42] Lung epithelium, [26,31,44,45] hepatocytes, islet beta cells Endothelins ECs Lung, [46] neural cells EG-VEGF ECs Endocrine glands [32] Brain-derived neurotrophic factor Brain microvascular endothelium Neuronal precursors, [33] islet endothelium Pigment epithelium-derived factor Brain capillaries Neural stem cells [35,47] Vessel-derived stromal-derived factor 1 (SDF-1/CXCL12) Microvascular endothelium Endoderm and pancreatic beta cells [48] Wnt2 Sinusoidal ECs Hepatocytes [26] S1P Plasma (plateles) Pancreatic multipotent progenitor cells [38] CTGF Pancreas capillaries Pancreatic endocrine cells [49] Laminin Islet capillaries Islet endocrine cells [50] Collagen IV Islet capillaries Islet beta-cells [51] BMP-2 ECs/MSCs Islet beta-cells [52,53] BMP-4 ECs/MSCs Islet beta-cells; hepatocytes; cardiomyocytes [52,53] BMPR1A ECs/MSCs Islet beta-cells; hepatocytes; cardiomyocytes [52,53] ECs: Endothelial cells; BMP: Bone morphogenetic protein; VEGF: Vascular endothelial cell growth factor; HGF: Hepatocyte growth factor; bFGF: Basic fibroblast growth factor; PDGF: Platelet-derived growth factor; CTGF: Connective tissue growth factor.

the notochord and factors such as fibroblast growth factor-2 (FGF-2) and activin-βB suppress the expression of sonic hedgehog (SHH) locally and promotes [60-63] expression of PDX-1 in the subjacent endoderm (Figure 1). Once the mesoderm layer starts proliferating, other signals from aortic endothelial cells (AECs) and mesen­ chyme (MCs) continue interacting with these PDX-1 expressing cells that give rise to acinar cells that harbor [64] pancreatic exocrine and endocrine progenitors (Figure 2). As these progenitors continue receiving more surrounding instructive signals, definition of cell function is established and the exocrine cells form acini while the [54] endocrine cells form islets of Langerhans (Figure 3). However, these islets contain immature beta cells that will become more mature after islet vascularization that allows closer interaction between beta cells and ECs.

N

FGF-2

Activin-bB

FG

PDX-1+/shh-

Figure 1 Early cell-cell interactions that give rise to pancreatic cells derived from definitive endoderm of the foregut (FG). Factors released by notochord (N) such as activin-βB and FGF-2 permit expression of PDX-1 and suppress expression of sonic hedgehog. FGF: Fibroblast growth factor; PDX: Pancreatic and duodenal homeobox.

Embryonic endothelium and endoderm pre-patterning

complete differentiation and maturation of pancreatic endocrine cells.

The vascular system is one of the first tissues that develop during embryogenesis. Mesodermal progenitors coalesce in the yolk sac and give rise to endothelium and [15] blood cells . Endothelial cells exert inductive effects on specific points were they are in contact with pre[20] patterned definitive endoderm (DE) cells of the FG . DE forms during gastrulation and Nodal, a member of transforming growth factor β (TGFβ) family, plays [65] a central role in DE formation . At these points of effective cell-cell interactions, the gut endoderm has to be competent to respond to EC-derived signals. Competence of these cells takes place during gastrulation when the mesoderm germ layer invades the middle [66] area between primitive ectoderm and endoderm .

ENDOTHELIUM ROLE IN THE PANCREATIC NICHE The pancreas originates from ventral and dorsal buds formed in the foregut at 8.5 d post-coitum (d.p.c) of gestation in mice and Carnegie stage 12 (CS12) [54-56] in humans . The cells that composed these buds express transcription factors such as PDX-1 which is a [57-59] key regulator of pancreas development . However, before these cells express these genes, the cells interact with other surrounding cells such as those that compose

WJEM|www.wjgnet.com

42

May 20, 2015|Volume 5|Issue 2|

Talavera-Adame D et al . Endothelium role in pancreas organogenesis AECs N

Collagen Ⅳ Laminin Ⅰ MCs

AECs

IECs BCs

?

?

DPB DPB

PDX-1+/insulin+

FG PDX-1+/shhFG

Figure 2 Endothelial-derived signals maintain expression of PDX-1 and promote pancreatic endocrine differentiation. Once the aorta is forming, aortic ECs (AECs) interact closely with FG/DE cells that maintain expression of PDX-1 and form the dorsal pancreatic bud (DPB). ECs: Endothelial cells; DE: Definitive endoderm; FG: Foregut; PDX: Pancreatic and duodenal homeobox.

Figure 3 Endothelial-derived signals promote the survival of pancreatic mesenchyme which is essential for pancreas development. Mesenchymal cells (MCs) appear between AECs and the dorsal pancreatic bud (DPB) and promote proliferation and survival of differentiated cells. Immature beta cells (BCs) that co-express PDX-1 and insulin migrate toward the mesenchyme and form cell clusters that will become islet of Langerhans that will recruit ECs that become islet ECs (iECs) and produce collagen IV and laminins which promote insulin expression. AECs crosstalk with MCs and maintain the integrity of these cells toward adequate exocrine and endocrine pancreas development. AECs: Aortic ECs; ECs: Endothelial cells; PDX: Pancreatic and duodenal homeobox.

Apparently mesoderm-derived cells are required to maintain the phenotype of posterior endoderm that includes the site where pancreas and duodenum will be formed. Therefore, anterior-posterior (A-P) endoderm axis will be sustained by the presence of mesodermderived factors such as Wingless-type MMTV integration site family (Wnt), fibroblast growth factor (FGF), bone morphogenetic proteins (BMPs), and retinoic acid [63,67] (RA) . For instance, high signaling of the canonical WNT/β-catenin pathway promotes endoderm posterior pattern with foregut-derived structures such as pancreas [68] and liver . Interestingly it has been found that some of [13,42,53,69-71] these factors are produced by ECs .

leading to inhibition of factors such as SHH and promotion [60] of PDX-1 expression . However, these permissive signals are apparently replaced by instructive signals from the growing mesoderm and AECs that become in close proximity with pre-patterned endoderm. Interestingly, ECs not only exert these signals directly to the pre-patterned endoderm but also promote the survival of adjacent mesodermal cells that produced essential factors such [64] as Islet-1 (Isl1) . Homozygous mice lacking expression -/of Flk-1 (flk-1 ) had absence of aorta with no formation of dorsal mesenchyme that led to diminished expression [64] of PDX-1 in the subjacent endoderm . However, lateral and ventral mesenchyme were not affected and PDX-1 [64] positive cells also appeared in the ventral endoderm . These findings indicate that aortic EC signaling is essential to maintain the dorsal mesenchyme and therefore to direct differentiation of dorsal pancreatic endoderm. -/Additionally, at later embryo stage, flk-1 mice showed dorsal mesenchyme that does not express Isl-1 in the absence of aortic ECs suggesting that endothelial-cell signaling promotes Isl-1 cell expression from dorsal [64] mesenchymal cells . In addition, it has been reported that mesenchymal cells also express bone morphogenetic proteins (BMPs) and that these proteins have a pivotal role [71,72] in pancreas development .

Pancreatic specification induced by surrounding endothelium

Endothelial signaling is required to induce insulin gene [20] expression during pancreas development . Cell-cell interactions between definitive endoderm and aortic endothelial cells take place at about 9-10 d.p.c. in mice [18,20] and give rise to PDX-1 expressing cells . Apparently, these interactions are also essential to promote insulin [20] expression in pre-patterned endoderm . However, signals from the developing pancreas to embryonic endothelium also promote endothelium-specific phenotype and these interactions are crucial for adequate [32] organ function in adulthood . As mentioned above, the first signals to promote expression of PDX-1 come from the notochord that produce factors such as activin-βB [60] and FGF-2 . These cross-talk take place between cells from notochord and cells from the subjacent endoderm

WJEM|www.wjgnet.com

43

May 20, 2015|Volume 5|Issue 2|

Talavera-Adame D et al . Endothelium role in pancreas organogenesis

Signals from pancreatic mesenchyme toward exocrine pancreas

that migrate are pancreatic endocrine progenitors that give rise islets of Langerhans composed of alpha, beta, delta, and PP cells that will produce glucagon, insulin, somatostatin, and pancreatic polypeptide respectively. These islets will be distributed differently into the pancreas and apparently will be subjected to different [74,75] stimuli . Although endocrine specification takes place before migration, maturation of endocrine cells occurs [54,76] at islet level and coincides with islet vascularization . There is a significant growth of islet cells that correlates with islet endothelial-cell proliferation in rats the first [76] week after birth . For instance, endocrine cells with [76] higher proliferative capacity closer to blood vessels . Furthermore, it is known that ECs are able to produce HGF which is a potent mitogen for beta cells and [31,77] ECs . Therefore, the endothelial signaling is essential for beta-cell maturation. For instance, immature beta cells are formed some days before birth and [76] maturation occurs several days after birth . During this period, immature beta cells express VEGF and start recruiting ECs to the vicinity of the islet to provide [32,78] signals for further differentiation and maturation . However, along with EC stimuli, another signals should also be considered. For example, hormones that can reach beta cells through the blood stream once the vascular network is established may have a role in cell [79] maturation . Apparently, endogenous insulin has a [80] minor role for the glucose homeostasis before birth . In this condition, insulin provided by the mother [80] regulates glucose in the fetus . This fact suggests that fetal beta cells are not mature enough at birth to maintain the glucose homeostasis and that further maturation can be promoted by ECs after birth.

It is well known that once the mesenchymal cells proliferate between aortic ECs and the foregut endoderm, the aorta is pulled out from the subjacent gut [20,55] (compare Figure 2 with Figure 3) . This fact implies that the subjacent foregut starts receiving signals from the mesenchyme and that a gradient can be formed with diluted signals from aortic ECs. However, it has been demonstrated that ECs maintain the integrity of the subjacent mesenchyme and that absence of aorta promotes apoptosis of the mesenchymal cells [64] and avoids the formation of the dorsal pancreas . As mentioned before, mesenchymal cells between aorta and foregut maintain the expression of PDX-1 in pancreatic endoderm cells through the expression of [54] the transcription factor Isl-1 . In addition, signals from mesenchyme such as Fgf-10 are essential to promote proliferation of pancreatic buds that already received [64] signals from ECs . Apparently, the specification of pancreatic fate is determined by permissive signals from notochord and instructive signals by ECs that are maintained by mesenchyme after the aorta is pulled out from the gut. It has been suggested recently that specification of endocrine phenotype also takes place during the close interaction between aortic ECs and pancreatic progenitors that express PDX-1 within the [54] pancreatic bud . Previous experiments indicated that insulin is expressing in foregut explants only after co[20] culturing with aortic endothelial cells . However, the signals from ECs that promote such specification in the foregut endoderm toward the formation of pancreatic endocrine progenitors are still under investigation. In the same way more characterization is required to identify the signals from ECs that promote survival and adequate function of mesenchyme. Therefore, functional mesenchyme is crucial for appropriate signaling to the subjacent foregut endoderm toward maintenance of the [43] pancreatic phenotype and branching .

ENDOTHELIUM ROLE IN PANCREATIC BETA-CELL FUNCTION The role of ECs in b-cell function and pathology has been [47,81-82] previously described . It has been found that ECs [83,84] from islets correspond to fenestrated endothelium . Apparently the characteristics of islet endothelium differ from pancreatic exocrine endothelium and endothelium [47,83,85] from other regions . For instance, pancreatic endocrine capillaries have higher diameter than exocrine capillaries and endothelium from endocrine capillaries have 10 times more fenestrae that endothelium from [83] exocrine capillaries . These facts suggest that cell-cell interactions and signaling between endothelium and the surrounding cells are different even in the same organ. Pancreatic beta cells have polarity with an apical and basolateral membrane and insulin vesicles are more [47,86] dense in the apical region close to ECs . This aspect is very important when considering the ability of beta cells to release insulin into the blood stream. After islet transplantation, suitable EC-signals for polarization are crucial for appropriate insulin release into the capillaries. Islet ECs express common markers of ECs but one specific marker called the proteinase inhibitor and

ENDOTHELIUM ROLE IN PANCREATIC BETA-CELL MATURATION Pancreatic endothelium during islet neogenesis

Pancreatic endocrine progenitors receive signals from aortic ECs and mesenchyme that determined their fate. However, they remain interspersed with the ductal cells that form the epithelium of the growing branches. At a certain time of development, these cells receive still unknown instructions to migrate from the ductal area to the mesenchyme region. Once in the mesenchyme, these pancreatic progenitors that are apart from ductal [73] cells form the islets of Langerhans . This fact raises two questions: (1) Are there signals from ECs that promote islet neogenesis? and (2) Once the blood vessels are formed are there factors in the blood stream that promote the final maturation of beta cells? The answer to these questions is still unknown. The cells

WJEM|www.wjgnet.com

44

May 20, 2015|Volume 5|Issue 2|

Talavera-Adame D et al . Endothelium role in pancreas organogenesis

A

B

25 mm

Figure 4 Analysis of blood vessels and insulin-producing cells in embryoid bodies obtained from human embryonic stem cell line H9. Human embryonic stem cells were cultured in suspension for 5 d to obtain embryoid bodies (EB). After attachment on coverslips for 24 h. Some EBs were cultured alone or together with human microvascular endothelial cells (HMECs). Then, after 20 d both groups of EBs were fixed and stained with with anti-proinsulin (green) (a marker for pancreatic beta cells), anti-CD31 (red) (a marker for endothelial cells), and DAPI (blue) (that stains the nuclei). A: EB cells cultured alone that do not show proinsulin or CD31 expression. In contrast with (B) EB cells co-cultured with HMECs at passage 14 in which we can find cells that express proinsulin in close proximity to cells that express CD31. HMECs did not stain positive for CD31 at the dilutions used indicating that the ECs are forming within EBs. [70,84]

[52,53,94]

angiostatin factor α1-antitrypsin . This marker of specific islet ECs can be absent in surrogate vasculature with deregulation of islet function. Apparently EPCs or islet ECs are important for islet revascularization after [87-89] transplantation . In this sense, any islet injury leads to islet restoration through recruitment of EPCs toward islet re-vascularization with specific islet ECs and beta cell [87] function . For instance, normoglycemia is improved in streptozotocin-treated animals after co-transplantation of [87] EPCs and islets .

mouse ESCs . These findings together with previous works pointed out the important role of ECs in beta-cell differentiation in vitro. We recently have observed that when human ESCs (cell line H9) are co-cultured with HMECs, the formation of proinsulin positive cells takes place in about twenty days in close proximity to internal ECs without the use of additional growth factors (Figure 4). In the model using mouse EBs, we observed that [53] ECs promote up-regulation of BMPs within EBs . However, the target cells that produce these BMPs are still unknown. One good candidate for these cells are mesenchymal cells since it has been demonstrated that ECs are essential to maintain dorsal pancreatic mesenchyme during pancreas morphogenesis that [64] may promote pancreas differentiation within EBs (Figure 5). However, further studies should be done to demonstrate that internal ECs are able to trigger beta cell differentiation through signaling to mesenchymal stem cells. At present, ECs can be generated in vitro from human induced pluripotential stem cells (hiPSCs) or human embryonic stem cells (hESCs) and this studies [95,96] can be important to answer this question . In this model, an excess of human microvascular ECs (HMECs) surround a human embryoid body which is composed of endoderm, ectoderm, and mesoderm cells. External ECs produce factors such as BMPs and other EC-derived factors that promote upregulation of endogenous BMPs in still unknown target cells (possibly mesenchymal or internal ECs). These BMPs together with other unknown factors may promote differentiation of multipotent cells (MC) toward beta-like cells (BC) and [52,53] other cell lineages (OCL) .

ENDOTHELIUM AND BETA-CELL REGENERATION IN VIVO AND IN VITRO As a first approach to investigate the role of endothelium in b-cell differentiation, we studied the role of in vivo surrogate vasculature in mouse embryoid body (EB) differentiation using quail chorioallantoic membranes [90] (CAM) . We found that some cells expressed cardiotin, myosin heavy chain, collagen Ⅳ, CD34, CD31, and neurofilament. Although some epithelial cells appeared, [90] no cells derived from endoderm were identified . Then, studies using co-cultures between human microvascular [52,53,91] ECs (HMECs) and mouse EBs were performed . In these studies, ECs from human dermis were able to induce differentiation of mouse EBs to pancreatic [52] progenitors and insulin-producing cells . Furthermore, BMP-2/-4 were involved in this differentiation process as evaluated by the effects of agonists (recombinant BMPs), and specific antagonists of BMP bioactivities (Noggin, [92] Chordin). BMPs are members of the TGF-β superfamily . In addition to the effects of BMP antagonists, we explored the levels of phosphorylation of SMAD1, 5, 8 in cells that [52,53] expressed proinsulin . The role of BMPs in pancreas [72,93] development has also been explored previously . We demonstrated that HMECs or mouse dermis as well as mouse AECs expressed BMPs and that BMP-2 and BMP-4 increased the phosphorylation levels of SMAD1,5,8 in pancreatic progenitors and beta-like cells derived from

WJEM|www.wjgnet.com

ENDOTHELIUM AND BETA-CELL PATHOGENESIS ECs play an important role for the pathogenesis of type 1 (T1DM) and type 2 diabetes mellitus (T2DM). ECs involvement in cellular diapedesis, inflammation,

45

May 20, 2015|Volume 5|Issue 2|

Talavera-Adame D et al . Endothelium role in pancreas organogenesis External ECs EB

2

?

3

BC Internal ECs or MCs

4

BM Ps

?

? BMPs MC

5

?

?

6

OCL

7 Figure 5 Diagram that explains the possible effects of external Endothelial cells toward in vitro beta-cell differentiation in human embryoid bodies. ?: Unknown factors, differentiation steps, or cell lineages; BMPs: Bone morphogenetic proteins; EC: Endothelial cells; EB: Embryoid bodies; BC: Beta cells; OCL: Other cells lineages; MC: Multipotent cell.

8 9

and Vessel fibrosis, which leads to islet dysfunction, has been demonstrated. In T1DM, Ecs allow infiltration [47] of leucocytes . Interestingly multiple diabetic comp­ lications relate to endothelial-cell function. The key to prevention or reversal of diabetes may rest in the recently emphasized role of endothelium in beta-cell [52,53] differentiation and maturation in vitro . In addition, it has been suggested that EC pathology can lead to islet dysfunction suggesting that ECs are [97] essential to maintain islet function in adults .

10

11

CONCLUSION

12

In this review we focused in the essential role of endothelium for pancreatic endocrine differentiation, functional maturation, and islet dysfunction. ECs play a key role during the differentiation of the dorsal pancreas by maintaining the expression of transcription factors necessary for pancreas development including endocrine progenitors. Before birth, immature beta cells recruit ECs close to their microenvironment and these ECs provide signals for further maturation and function of pancreatic beta cells. In addition, ECs co-transplanted with islets have demonstrated to improve the engraftment of human islets. Finally, ECs are able to provide signals in vitro for derivation of functional beta-like cells from human pluripotent stem cells. Therefore, the study of interactions between EC and beta cells is relevant for future clinical applications in regenerative medicine.

13 14 15 16 17

18 19

REFERENCES 1

Liew CG. Generation of insulin-producing cells from pluripotent

WJEM|www.wjgnet.com

20

46

stem cells: from the selection of cell sources to the optimization of protocols. Rev Diabet Stud 2010; 7: 82-92 [PMID: 21060967 DOI: 10.1900/RDS.2010.7.82] Pagliuca FW, Millman JR, Gürtler M, Segel M, Van Dervort A, Ryu JH, Peterson QP, Greiner D, Melton DA. Generation of functional human pancreatic β cells in vitro. Cell 2014; 159: 428-439 [PMID: 25303535 DOI: 10.1016/j.cell.2014.09.040] Lock LT, Tzanakakis ES. Stem/Progenitor cell sources of insulinproducing cells for the treatment of diabetes. Tissue Eng 2007; 13: 1399-1412 [PMID: 17550339 DOI: 10.1089/ten.2007.0047] Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science 1998; 282: 1145-1147 [PMID: 9804556 DOI: 10.1126/science.282.5391.1145] Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126: 663-676 [PMID: 16904174 DOI: 10.1016/ j.cell.2006.07.024] Segev H, Fishman B, Ziskind A, Shulman M, Itskovitz-Eldor J. Differentiation of human embryonic stem cells into insulinproducing clusters. Stem Cells 2004; 22: 265-274 [PMID: 15153604] Rezania A, Bruin JE, Riedel MJ, Mojibian M, Asadi A, Xu J, Gauvin R, Narayan K, Karanu F, O’Neil JJ, Ao Z, Warnock GL, Kieffer TJ. Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating preexisting diabetes in mice. Diabetes 2012; 61: 2016-2029 [PMID: 22740171 DOI: 10.2337/db11-1711] Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki KL, Tzukerman M. Insulin production by human embryonic stem cells. Diabetes 2001; 50: 1691-1697 [PMID: 11473026] Assady S. Challenges and prospects for stem cell-based therapy in diabetes mellitus. Isr Med Assoc J 2009; 11: 212-215 [PMID: 19603593] D’Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter MK, Baetge EE. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol 2006; 24: 1392-1401 [PMID: 17053790 DOI: 10.1038/nbt1259] Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H, Richardson M, Smart NG, Cunningham J, Agulnick AD, D’Amour KA, Carpenter MK, Baetge EE. Pancreatic endoderm derived from human embryonic stem cells generates glucoseresponsive insulin-secreting cells in vivo. Nat Biotechnol 2008; 26: 443-452 [PMID: 18288110 DOI: 10.1038/nbt1393] Coultas L, Chawengsaksophak K, Rossant J. Endothelial cells and VEGF in vascular development. Nature 2005; 438: 937-945 [PMID: 16355211] Herbert SP, Stainier DY. Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat Rev Mol Cell Biol 2011; 12: 551-564 [PMID: 21860391 DOI: 10.1038/nrm3176] Risau W. Differentiation of endothelium. FASEB J 1995; 9: 926-933 [PMID: 7615161] Noden DM. Embryonic origins and assembly of blood vessels. Am Rev Respir Dis 1989; 140: 1097-1103 [PMID: 2478056 DOI: 10.1164/ajrccm/140.4.1097] Nikolova G, Strilic B, Lammert E. The vascular niche and its basement membrane. Trends Cell Biol 2007; 17: 19-25 [PMID: 17129728 DOI: 10.1016/j.tcb.2006.11.005] Matsumoto K, Yoshitomi H, Rossant J, Zaret KS. Liver organogenesis promoted by endothelial cells prior to vascular function. Science 2001; 294: 559-563 [PMID: 11577199 DOI: 10.1126/science.1063889] Nikolova G, Lammert E. Interdependent development of blood vessels and organs. Cell Tissue Res 2003; 314: 33-42 [PMID: 12898210 DOI: 10.1007/s00441-003-0739-8] Crivellato E, Nico B, Ribatti D. Contribution of endothelial cells to organogenesis: a modern reappraisal of an old Aristotelian concept. J Anat 2007; 211: 415-427 [PMID: 17683480 DOI: 10.1111/ j.1469-7580.2007.00790.x] Lammert E, Cleaver O, Melton D. Induction of pancreatic

May 20, 2015|Volume 5|Issue 2|

Talavera-Adame D et al . Endothelium role in pancreas organogenesis

21 22 23

24 25

26

27

28 29

30

31

32 33

34

35

36 37

differentiation by signals from blood vessels. Science 2001; 294: 564-567 [PMID: 11577200 DOI: 10.1126/science.1064344] Lammert E, Cleaver O, Melton D. Role of endothelial cells in early pancreas and liver development. Mech Dev 2003; 120: 59-64 [PMID: 12490296] Mantovani A, Bussolino F, Dejana E. Cytokine regulation of endothelial cell function. FASEB J 1992; 6: 2591-2599 [PMID: 1592209] Orr FW, Wang HH, Lafrenie RM, Scherbarth S, Nance DM. Interactions between cancer cells and the endothelium in metastasis. J Pathol 2000; 190: 310-329 [PMID: 10685065 DOI: 10.1002/(SIC I)1096-9896(200002)190: 33.0.CO; 2-P] Crivellato E. The role of angiogenic growth factors in organogenesis. Int J Dev Biol 2011; 55: 365-375 [PMID: 21858761 DOI: 10.1387/ ijdb.103214ec] Butler JM, Nolan DJ, Vertes EL, Varnum-Finney B, Kobayashi H, Hooper AT, Seandel M, Shido K, White IA, Kobayashi M, Witte L, May C, Shawber C, Kimura Y, Kitajewski J, Rosenwaks Z, Bernstein ID, Rafii S. Endothelial cells are essential for the selfrenewal and repopulation of Notch-dependent hematopoietic stem cells. Cell Stem Cell 2010; 6: 251-264 [PMID: 20207228 DOI: 10.1016/j.stem.2010.02.001] Ding BS, Nolan DJ, Butler JM, James D, Babazadeh AO, Rosenwaks Z, Mittal V, Kobayashi H, Shido K, Lyden D, Sato TN, Rabbany SY, Rafii S. Inductive angiocrine signals from sinusoidal endothelium are required for liver regeneration. Nature 2010; 468: 310-315 [PMID: 21068842 DOI: 10.1038/nature09493.Inductive] Brantley-Sieders DM, Dunaway CM, Rao M, Short S, Hwang Y, Gao Y, Li D, Jiang A, Shyr Y, Wu JY, Chen J. Angiocrine factors modulate tumor proliferation and motility through EphA2 repression of Slit2 tumor suppressor function in endothelium. Cancer Res 2011; 71: 976-987 [PMID: 21148069 DOI: 10.1158/0008-5472. CAN-10-3396] Bahary N, Zon LI. Development. Endothelium--chicken soup for the endoderm. Science 2001; 294: 530-531 [PMID: 11577202 DOI: 10.1126/science.1066282] Uchimura T, Komatsu Y, Tanaka M, McCann KL, Mishina Y. Bmp2 and Bmp4 genetically interact to support multiple aspects of mouse development including functional heart development. Genesis 2009; 47: 374-384 [PMID: 19391114 DOI: 10.1002/ dvg.20511] Takahashi T, Huynh-Do U, Daniel TO. Renal microvascular assembly and repair: power and promise of molecular definition. Kidney Int 1998; 53: 826-835 [PMID: 9551388 DOI: 10.1111/ j.1523-1755.1998.00822.x] LeCouter J, Moritz DR, Li B, Phillips GL, Liang XH, Gerber HP, Hillan KJ, Ferrara N. Angiogenesis-independent endothelial protection of liver: role of VEGFR-1. Science 2003; 299: 890-893 [PMID: 12574630 DOI: 10.1126/science.1079562] Cleaver O, Melton DA. Endothelial signaling during development. Nat Med 2003; 9: 661-668 [PMID: 12778164 DOI: 10.1038/ nm0603-661] Palmer TD, Willhoite AR, Gage FH. Vascular niche for adult hippocampal neurogenesis. J Comp Neurol 2000; 425: 479-494 [PMID: 10975875 DOI: 10.1002/1096-9861(20001002)425: 43.0.CO; 2-3] Louissaint A, Rao S, Leventhal C, Goldman SA. Coordinated interaction of neurogenesis and angiogenesis in the adult songbird brain. Neuron 2002; 34: 945-960 [PMID: 12086642 DOI: 10.1016/ S0896-6273(02)00722-5] Ramírez-Castillejo C, Sánchez-Sánchez F, Andreu-Agulló C, Ferrón SR, Aroca-Aguilar JD, Sánchez P, Mira H, Escribano J, Fariñas I. Pigment epithelium-derived factor is a niche signal for neural stem cell renewal. Nat Neurosci 2006; 9: 331-339 [PMID: 16491078 DOI: 10.1038/nn1657] Katsumoto K, Kume S. The role of CXCL12-CXCR4 signaling pathway in pancreatic development. Theranostics 2013; 3: 11-17 [PMID: 23382781 DOI: 10.7150/thno.4806] Kumar M, Jordan N, Melton D, Grapin-Botton A. Signals from lateral plate mesoderm instruct endoderm toward a pancreatic fate.

WJEM|www.wjgnet.com

38

39

40

41

42 43

44

45

46 47

48

49

50

51

52

53

47

Dev Biol 2003; 259: 109-122 [PMID: 12812792 DOI: 10.1016/ S0012-1606(03)00183-0] Edsbagge J, Johansson JK, Esni F, Luo Y, Radice GL, Semb H. Vascular function and sphingosine-1-phosphate regulate development of the dorsal pancreatic mesenchyme. Development 2005; 132: 1085-1092 [PMID: 15689381 DOI: 10.1242/dev.01643] Heinis M, Simon MT, Ilc K, Mazure NM, Pouysségur J, Scharfmann R, Duvillié B. Oxygen tension regulates pancreatic beta-cell differentiation through hypoxia-inducible factor 1alpha. Diabetes 2010; 59: 662-669 [PMID: 20009089 DOI: 10.2337/ db09-0891] Kopp HG, Avecilla ST, Hooper AT, Rafii S. The bone marrow vascular niche: home of HSC differentiation and mobilization. Physiology (Bethesda) 2005; 20: 349-356 [PMID: 16174874 DOI: 10.1152/physiol.00025.2005] Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N, Vincent P, Pumiglia K, Temple S. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science 2004; 304: 1338-1340 [PMID: 15060285 DOI: 10.1126/ science.1095505] Muñoz-Chápuli R, Quesada AR, Angel Medina M. Angiogenesis and signal transduction in endothelial cells. Cell Mol Life Sci 2004; 61: 2224-2243 [PMID: 15338053 DOI: 10.1007/s00018-004-4070-7] Glienke J, Schmitt AO, Pilarsky C, Hinzmann B, Weiss B, Rosenthal A, Thierauch KH. Differential gene expression by endothelial cells in distinct angiogenic states. Eur J Biochem 2000; 267: 2820-2830 [PMID: 10785405] Yamamoto H, Yun EJ, Gerber HP, Ferrara N, Whitsett JA, Vu TH. Epithelial-vascular cross talk mediated by VEGF-A and HGF signaling directs primary septae formation during distal lung morphogenesis. Dev Biol 2007; 308: 44-53 [PMID: 17583691 DOI: 10.1016/j.ydbio.2007.04.042] LeCouter J, Lin R, Ferrara N. Endocrine gland-derived VEGF and the emerging hypothesis of organ-specific regulation of angiogenesis. Nat Med 2002; 8: 913-917 [PMID: 12205443 DOI: 10.1038/nm0902-913] Masaki T. Historical review: Endothelin. Trends Pharmacol Sci 2004; 25: 219-224 [PMID: 15063086 DOI: 10.1016/j.tips.2004.02.008] Olsson R, Carlsson PO. The pancreatic islet endothelial cell: emerging roles in islet function and disease. Int J Biochem Cell Biol 2006; 38: 710-714 [PMID: 16607697 DOI: 10.1016/ j.biocel.2006.02.004] Yano T, Liu Z, Donovan J, Thomas MK, Habener JF. Stromal cell derived factor-1 (SDF-1)/CXCL12 attenuates diabetes in mice and promotes pancreatic beta-cell survival by activation of the prosurvival kinase Akt. Diabetes 2007; 56: 2946-2957 [PMID: 17878289 DOI: 10.2337/db07-0291] Crawford LA, Guney MA, Oh YA, Deyoung RA, Valenzuela DM, Murphy AJ, Yancopoulos GD, Lyons KM, Brigstock DR, Economides A, Gannon M. Connective tissue growth factor (CTGF) inactivation leads to defects in islet cell lineage allocation and betacell proliferation during embryogenesis. Mol Endocrinol 2009; 23: 324-336 [PMID: 19131512 DOI: 10.1210/me.2008-0045] Nikolova G, Jabs N, Konstantinova I, Domogatskaya A, Tryggvason K, Sorokin L, Fässler R, Gu G, Gerber HP, Ferrara N, Melton DA, Lammert E. The vascular basement membrane: a niche for insulin gene expression and Beta cell proliferation. Dev Cell 2006; 10: 397-405 [PMID: 16516842 DOI: 10.1016/j.devcel.2006.01.015] Kaido T, Yebra M, Cirulli V, Montgomery AM. Regulation of human beta-cell adhesion, motility, and insulin secretion by collagen IV and its receptor alpha1beta1. J Biol Chem 2004; 279: 53762-53769 [PMID: 15485856 DOI: 10.1074/jbc.M411202200] Talavera-Adame D, Wu G, He Y, Ng TT, Gupta A, Kurtovic S, Hwang JY, Farkas DL, Dafoe DC. Endothelial cells in coculture enhance embryonic stem cell differentiation to pancreatic progenitors and insulin-producing cells through BMP signaling. Stem Cell Rev 2011; 7: 532-543 [PMID: 21298405 DOI: 10.1007/ s12015-011-9232-z] Talavera-Adame D, Gupta A, Kurtovic S, Chaiboonma KL, Arumugaswami V, Dafoe DC. Bone morphogenetic protein-2/-4

May 20, 2015|Volume 5|Issue 2|

Talavera-Adame D et al . Endothelium role in pancreas organogenesis

54 55

56 57

58

59 60 61 62 63

64

65

66 67

68

69

70

71

72

upregulation promoted by endothelial cells in coculture enhances mouse embryoid body differentiation. Stem Cells Dev 2013; 22: 3252-3260 [PMID: 23924071 DOI: 10.1089/scd.2013.0013] Habener JF, Kemp DM, Thomas MK. Minireview: transcriptional regulation in pancreatic development. Endocrinology 2005; 146: 1025-1034 [PMID: 15604203 DOI: 10.1210/en.2004-1576] Jennings RE, Berry AA, Kirkwood-Wilson R, Roberts NA, Hearn T, Salisbury RJ, Blaylock J, Piper Hanley K, Hanley NA. Development of the human pancreas from foregut to endocrine commitment. Diabetes 2013; 62: 3514-3522 [PMID: 23630303 DOI: 10.2337/db12-1479] Hill MA. Early human development. Clin Obstet Gynecol 2007; 50: 2-9 [PMID: 17304021] Grapin-Botton A. Antero-posterior patterning of the vertebrate digestive tract: 40 years after Nicole Le Douarin’s PhD thesis. Int J Dev Biol 2005; 49: 335-347 [PMID: 15906249 DOI: 10.1387/ ijdb.041946ag] Gao N, LeLay J, Vatamaniuk MZ, Rieck S, Friedman JR, Kaestner KH. Dynamic regulation of Pdx1 enhancers by Foxa1 and Foxa2 is essential for pancreas development. Genes Dev 2008; 22: 3435-3448 [PMID: 19141476 DOI: 10.1101/gad.1752608.lineages] Jensen J. Gene regulatory factors in pancreatic development. Dev Dyn 2004; 229: 176-200 [PMID: 14699589 DOI: 10.1002/ dvdy.10460] Hebrok M, Kim SK, Melton DA. Notochord repression of endodermal Sonic hedgehog permits pancreas development. Genes Dev 1998; 12: 1705-1713 [PMID: 9620856] Stemple DL. Structure and function of the notochord: an essential organ for chordate development. Development 2005; 132: 2503-2512 [PMID: 15890825 DOI: 10.1242/dev.01812] Kim SK, Hebrok M. Intercellular signals regulating pancreas development and function. Genes Dev 2001; 15: 111-127 [PMID: 11157769 DOI: 10.1101/gad.859401] Ameri J, Ståhlberg A, Pedersen J, Johansson JK, Johannesson MM, Artner I, Semb H. FGF2 specifies hESC-derived definitive endoderm into foregut/midgut cell lineages in a concentrationdependent manner. Stem Cells 2010; 28: 45-56 [PMID: 19890880 DOI: 10.1002/stem.249] Jacquemin P, Yoshitomi H, Kashima Y, Rousseau GG, Lemaigre FP, Zaret KS. An endothelial-mesenchymal relay pathway regulates early phases of pancreas development. Dev Biol 2006; 290: 189-199 [PMID: 16386727 DOI: 10.1016/j.ydbio.2005.11.023] Vincent SD, Dunn NR, Hayashi S, Norris DP, Robertson EJ. Cell fate decisions within the mouse organizer are governed by graded Nodal signals. Genes Dev 2003; 17: 1646-1662 [PMID: 12842913 DOI: 10.1101/gad.1100503] McCracken KW, Wells JM. Molecular pathways controlling pancreas induction. Semin Cell Dev Biol 2012; 23: 656-662 [PMID: 22743233 DOI: 10.1016/j.semcdb.2012.06.009] Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, Hoskins EE, Kalinichenko VV, Wells SI, Zorn AM, Shroyer NF, Wells JM. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 2011; 470: 105-109 [PMID: 21151107 DOI: 10.1038/nature09691] McLin VA, Rankin SA, Zorn AM. Repression of Wnt/betacatenin signaling in the anterior endoderm is essential for liver and pancreas development. Development 2007; 134: 2207-2217 [PMID: 17507400 DOI: 10.1242/dev.001230] Lai L, Bohnsack BL, Niederreither K, Hirschi KK. Retinoic acid regulates endothelial cell proliferation during vasculogenesis. Development 2003; 130: 6465-6474 [PMID: 14627725 DOI: 10.1242/dev.00887] Linn T, Schneider K, Hammes HP, Preissner KT, Brandhorst H, Morgenstern E, Kiefer F, Bretzel RG. Angiogenic capacity of endothelial cells in islets of Langerhans. FASEB J 2003; 17: 881-883 [PMID: 12670881] Talavera-Adame D, Xiong Y, Zhao T, Arias AE, Sierra-Honigmann MR, Farkas DL. Quantitative and morphometric evaluation of the angiogenic effects of leptin. J Biomed Opt 2008; 13: 064017 [PMID: 19123663 DOI: 10.1117/1.3028010]

WJEM|www.wjgnet.com

73 74

75

76

77

78

79

80

81

82

83 84

85

86 87

88

48

Ahnfelt-Rønne J, Ravassard P, Pardanaud-Glavieux C, Scharfmann R, Serup P. Mesenchymal bone morphogenetic protein signaling is required for normal pancreas development. Diabetes 2010; 59: 1948-1956 [PMID: 20522595 DOI: 10.2337/db09-1010] Paris M, Tourrel-Cuzin C, Plachot C, Ktorza A. Review: pancreatic beta-cell neogenesis revisited. Exp Diabesity Res 2014; 5: 111-121 [PMID: 15203882 DOI: 10.1080/15438600490455079] Bosco D, Armanet M, Morel P, Niclauss N, Sgroi A, Muller YD, Giovannoni L, Parnaud G, Berney T. Unique arrangement of alphaand beta-cells in human islets of Langerhans. Diabetes 2010; 59: 1202-1210 [PMID: 20185817 DOI: 10.2337/db09-1177] Wang X, Misawa R, Zielinski MC, Cowen P, Jo J, Periwal V, Ricordi C, Khan A, Szust J, Shen J, Millis JM, Witkowski P, Hara M. Regional differences in islet distribution in the human pancreas-preferential beta-cell loss in the head region in patients with type 2 diabetes. PLoS One 2013; 8: e67454 [PMID: 23826303 DOI: 10.1371/journal.pone.0067454] Johansson M, Andersson A, Carlsson PO, Jansson L. Perinatal development of the pancreatic islet microvasculature in rats. J Anat 2006; 208: 191-196 [PMID: 16441563 DOI: 10.1111/ j.1469-7580.2006.00520.x] Otonkoski T, Cirulli V, Beattie M, Mally MI, Soto G, Rubin JS, Hayek A. A role for hepatocyte growth factor/scatter factor in fetal mesenchyme-induced pancreatic beta-cell growth. Endocrinology 1996; 137: 3131-3139 [PMID: 8770939] Inoue M, Hager JH, Ferrara N, Gerber HP, Hanahan D. VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesis. Cancer Cell 2002; 1: 193-202 [PMID: 12086877] Hardikar AA, Wang XY, Williams LJ, Kwok J, Wong R, Yao M, Tuch BE. Functional maturation of fetal porcine beta-cells by glucagon-like peptide 1 and cholecystokinin. Endocrinology 2002; 143: 3505-3514 [PMID: 12193564 DOI: 10.1210/en.2001-211344] Duvillié B, Cordonnier N, Deltour L, Dandoy-Dron F, Itier JM, Monthioux E, Jami J, Joshi RL, Bucchini D. Phenotypic alterations in insulin-deficient mutant mice. Proc Natl Acad Sci USA 1997; 94: 5137-5140 [PMID: 9144203] Richards OC, Raines SM, Attie AD. The role of blood vessels, endothelial cells, and vascular pericytes in insulin secretion and peripheral insulin action. Endocr Rev 2010; 31: 343-363 [PMID: 20164242 DOI: 10.1210/er.2009-0035] Sabra G, Vermette P. A 3D cell culture system: separation distance between INS-1 cell and endothelial cell monolayers co-cultured in fibrin influences INS-1 cells insulin secretion. Biotechnol Bioeng 2013; 110: 619-627 [PMID: 22949028 DOI: 10.1002/bit.24716] Henderson JR, Moss MC. A morphometric study of the endocrine and exocrine capillaries of the pancreas. Q J Exp Physiol 1985; 70: 347-356 [PMID: 3898188] Lou J, Triponez F, Oberholzer J, Wang H, Yu D, Buhler L, Cretin N, Mentha G, Wollheim CB, Morel P. Expression of alpha-1 proteinase inhibitor in human islet microvascular endothelial cells. Diabetes 1999; 48: 1773-1778 [PMID: 10480607] Mattsson G, Danielsson A, Kriz V, Carlsson PO, Jansson L. Endothelial cells in endogenous and transplanted pancreatic islets: differences in the expression of angiogenic peptides and receptors. Pancreatology 2006; 6: 86-95 [PMID: 16327285] Bonner-Weir S. Morphological evidence for pancreatic polarity of beta-cell within islets of Langerhans. Diabetes 1988; 37: 616-621 [PMID: 3282948 DOI: 10.2337/diabetes.37.5.616] Quaranta P, Antonini S, Spiga S, Mazzanti B, Curcio M, Mulas G, Diana M, Marzola P, Mosca F, Longoni B. Co-transplantation of endothelial progenitor cells and pancreatic islets to induce longlasting normoglycemia in streptozotocin-treated diabetic rats. PLoS One 2014; 9: e94783 [PMID: 24733186 DOI: 10.1371/journal. pone.0094783] Brissova M, Fowler M, Wiebe P, Shostak A, Shiota M, Radhika A, Lin PC, Gannon M, Powers AC. Intraislet endothelial cells contribute to revascularization of transplanted pancreatic islets. Diabetes 2004; 53: 1318-1325 [PMID: 15111502 DOI: 10.2337/ diabetes.53.5.1318]

May 20, 2015|Volume 5|Issue 2|

Talavera-Adame D et al . Endothelium role in pancreas organogenesis 89

90

91

92 93

Hess D, Li L, Martin M, Sakano S, Hill D, Strutt B, Thyssen S, Gray DA, Bhatia M. Bone marrow-derived stem cells initiate pancreatic regeneration. Nat Biotechnol 2003; 21: 763-770 [PMID: 12819790 DOI: 10.1038/nbt841] Talavera-Adame D, Dafoe DC, Ng TT, Wachsmann-Hogiu S, Castillo-Henkel C, Farkas DL. Enhancement of embryonic stem cell differentiation promoted by avian chorioallantoic membranes. Tissue Eng Part A 2009; 15: 3193-3200 [PMID: 19364272 DOI: 10.1089/ten.TEA.2009.0024] Banerjee I, Sharma N, Yarmush M. Impact of co-culture on pancreatic differentiation of embryonic stem cells. J Tissue Eng Regen Med 2011; 5: 313-323 [PMID: 20717889 DOI: 10.1002/ term.317] Bragdon B, Moseychuk O, Saldanha S, King D, Julian J, Nohe A. Bone morphogenetic proteins: a critical review. Cell Signal 2011; 23: 609-620 [PMID: 20959140 DOI: 10.1016/j.cellsig.2010.10.003] Goulley J, Dahl U, Baeza N, Mishina Y, Edlund H. BMP4BMPR1A signaling in beta cells is required for and augments glucose-stimulated insulin secretion. Cell Metab 2007; 5: 207-219

94

95

96

97

[PMID: 17339028 DOI: 10.1016/j.cmet.2007.01.009] Talavera-Adame D, Ng TT, Gupta A, Kurtovic S, Wu GD, Dafoe DC. Characterization of microvascular endothelial cells isolated from the dermis of adult mouse tails. Microvasc Res 2011; 82: 97-104 [PMID: 21570988 DOI: 10.1016/j.mvr.2011.04.009] Wang ZZ, Au P, Chen T, Shao Y, Daheron LM, Bai H, Arzigian M, Fukumura D, Jain RK, Scadden DT. Endothelial cells derived from human embryonic stem cells form durable blood vessels in vivo. Nat Biotechnol 2007; 25: 317-318 [PMID: 17322871 DOI: 10.1038/nbt1287] Kurtovic S, Ng TT, Gupta A, Arumugaswami V, Chaiboonma KL, Aminzadeh MA, Makkar R, Dafoe DC, Talavera-Adame D. Leptin enhances endothelial cell differentiation and angiogenesis in murine embryonic stem cells. Microvasc Res 2015; 97: 65-74 [PMID: 25250519 DOI: 10.1016/j.mvr.2014.09.004] Almaça J, Molina J, Arrojo E Drigo R, Abdulreda MH, Jeon WB, Berggren PO, Caicedo A, Nam HG. Young capillary vessels rejuvenate aged pancreatic islets. Proc Natl Acad Sci USA 2014; 111: 17612-17617 [PMID: 25404292 DOI: 10.1073/pnas.1414053111]

P- Reviewer: Sanal MG, Sakata N

WJEM|www.wjgnet.com

49

S- Editor: Ji FF L- Editor: A E- Editor: Lu YJ

May 20, 2015|Volume 5|Issue 2|

Published by Baishideng Publishing Group Inc 8226 Regency Drive, Pleasanton, CA 94588, USA Telephone: +1-925-223-8242 Fax: +1-925-223-8243 E-mail: [email protected] Help Desk: http://www.wjgnet.com/esps/helpdesk.aspx http://www.wjgnet.com

© 2015 Baishideng Publishing Group Inc. All rights reserved.

Endothelium-derived essential signals involved in pancreas organogenesis.

Endothelial cells (ECs) are essential for pancreas differentiation, endocrine specification, and endocrine function. They are also involved in the phy...
2MB Sizes 1 Downloads 7 Views